Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 20(20): e2308680, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38225709

RESUMEN

Gut microbiota function has numerous effects on humans and the diet humans consume has emerged as a pivotal determinant of gut microbiota function. Here, a new concept that gut microbiota can be trained by diet-derived exosome-like nanoparticles (ELNs) to release healthy outer membrane vesicles (OMVs) is introduced. Specifically, OMVs released from garlic ELN (GaELNs) trained human gut Akkermansia muciniphila (A. muciniphila) can reverse high-fat diet-induced type 2 diabetes (T2DM) in mice. Oral administration of OMVs released from GaELNs trained A. muciniphila can traffick to the brain where they are taken up by microglial cells, resulting in inhibition of high-fat diet-induced brain inflammation. GaELNs treatment increases the levels of OMV Amuc-1100, P9, and phosphatidylcholines. Increasing the levels of Amuc-1100 and P9 leads to increasing the GLP-1 plasma level. Increasing the levels of phosphatidylcholines is required for inhibition of cGas and STING-mediated inflammation and GLP-1R crosstalk with the insulin pathway that leads to increasing expression of Insulin Receptor Substrate (IRS1 and IRS2) on OMV targeted cells. These findings reveal a molecular mechanism whereby OMVs from plant nanoparticle-trained gut bacteria regulate genes expressed in the brain, and have implications for the treatment of brain dysfunction caused by a metabolic syndrome.


Asunto(s)
Eje Cerebro-Intestino , Diabetes Mellitus Tipo 2 , Exosomas , Ajo , Microbioma Gastrointestinal , Nanopartículas , Diabetes Mellitus Tipo 2/metabolismo , Ajo/química , Animales , Nanopartículas/química , Exosomas/metabolismo , Ratones , Akkermansia , Humanos , Masculino , Dieta Alta en Grasa , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/patología
2.
Microb Pathog ; 188: 106562, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38307370

RESUMEN

Bacterial outer membrane vesicles (OMVs) can package and deliver virulence factors into host cells, which is an important mechanism mediating host-pathogen interactions. It has been reported that small RNAs (sRNAs) can be packed into OMVs with varying relative abundance, which might affect the function and/or stability of host mRNAs. In this study, we used OptiPrep density gradient ultra-high-speed centrifugation to purify OMVs from Pseudomonas aeruginosa. Next, the sequences and abundance of sRNAs were detected by using Small RNA-Seq. In particular, sRNA4518698, sRNA2316613 and sRNA809738 were the three most abundant sRNAs in OMVs, which are all fragments of P. aeruginosa non-coding RNAs. sRNAs were shielded within the interior of OMVs and remained resistant to external RNase cleavage. The miRanda and RNAhybrid analysis demonstrated that those sRNAs could target a large number of host mRNAs, which were enriched in host immune responses by the functions of GO and KEGG enrichment. Experimentally, we demonstrated that the transfection of synthetic sRNA4518698, sRNA2316613, or sRNA809738 could reduce the expression of innate immune response genes in RAW264.7 cells. Together, we demonstrated that P. aeruginosa OMVs sRNAs can regulate innate immune responses. This study uncovered a mechanism in which the OMVs regulate host responses by transferring bacterial sRNAs.


Asunto(s)
Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/fisiología , Infecciones por Pseudomonas/microbiología , Inmunidad Innata , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Interacciones Huésped-Patógeno , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Helicobacter ; 29(1): e13031, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37997013

RESUMEN

BACKGROUND: Outer membrane vesicles (OMVs) are spontaneously released by Gram-negative bacteria and influence bacteria-host interactions by acting as a delivery system for bacterial components and by interacting directly with host cells. Helicobacter pylori, a pathogenic bacterium that chronically colonizes the human stomach, also sheds OMVs, and their impact on bacterial-mediated diseases is still being elucidated. MATERIALS AND METHODS: Transcriptomic profiling of the human gastric cell line MKN74 upon challenge with H. pylori OMVs compared to control and infected cells was performed using the Ion AmpliSeq™ Transcriptome Human Gene Expression Panel to understand the gene expression changes that human gastric epithelial cells might undergo when exposed to H. pylori OMVs. RESULTS: H. pylori OMVs per se modify the gene expression profile of gastric epithelial cells, adding another layer of (gene) regulation to the already complex host-bacteria interaction. The most enriched pathways include those related to amino acid metabolism, mitogen-activated protein kinase signaling, autophagy, and ferroptosis, whereas the cell cycle, DNA replication, and DNA repair were the most downregulated. The transcriptomic changes induced by OMVs were mostly similar to those induced by the parental bacteria, likely amplifying the effects of the bacterium itself. CONCLUSIONS: Our data provide a valuable portrayal of the transcriptomic remodeling of gastric cells induced by H. pylori OMVs. It demonstrates the breadth of cellular pathways and genes affected by OMVs, most previously unreported, which can be further dissected for the underlying molecular mediators and explored to understand the pathobiology of the full spectrum of H. pylori-mediated diseases.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Helicobacter pylori/fisiología , Transcriptoma , Infecciones por Helicobacter/microbiología , Estómago , Perfilación de la Expresión Génica
4.
Appl Microbiol Biotechnol ; 108(1): 29, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38159117

RESUMEN

Small noncoding RNAs (sncRNAs) play important regulatory roles in bacterial physiological processes and host-pathogen interactions. Meanwhile, bacterial outer membrane vesicles (OMVs), as naturally secreted outer membrane structures, play a vital role in the interaction between bacteria and their living environment, including the host environment. However, most current studies focus on the biological functions of sncRNAs in bacteria or hosts, while neglecting the roles and regulatory mechanisms of the OMVs that encapsulate these sncRNAs. Therefore, this review aims to summarize the intracellular regulatory roles of bacterial sncRNAs in promoting pathogen survival by regulating virulence, modulating bacterial drug resistance, and regulating iron metabolism, and their extracellular regulatory function for influencing host immunity through host-pathogen interactions. Additionally, we introduce the key role played by OMVs, which serve as important cargoes in bacterial sncRNA-host interactions. We propose emerging pathways of sncRNA action to further discuss the mode of host-pathogen interactions, highlighting that the inhibition of sncRNA delivery by OMVs may prevent the occurrence of infection to some extent. Hence, this review lays the foundation for future prophylactic treatments against bacterial infections and strategies for addressing bacterial drug resistance. KEY POINTS: •sncRNAs have intracellular and extracellular regulatory functions in bacterial physiological processes and host-pathogen interactions. •OMVs are potential mediators between bacterial sncRNAs and host cells. •OMVs encapsulating sncRNAs have more potential biological functions.


Asunto(s)
Vesículas Extracelulares , ARN Pequeño no Traducido , ARN Pequeño no Traducido/genética , Proteínas de la Membrana Bacteriana Externa/genética , Bacterias/metabolismo , Interacciones Huésped-Patógeno/fisiología , Interacciones Microbiota-Huesped , Vesículas Extracelulares/metabolismo
5.
Nano Lett ; 23(18): 8593-8601, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37625135

RESUMEN

Despite the great progress of current bacterially based biotherapeutics, their unsatisfying efficacy and underlying safety problems have limited their clinical application. Herein, inspired by probiotic Escherichia coli strain Nissle 1917, probiotic-derived outer membrane vesicles (OMVs) are found to serve as an effective therapeutic platform for the treatment of inflammatory bowel disease (IBD). To further enhance the therapeutic effect, the probiotic-derived OMV-encapsulating manganese dioxide nanozymes are constructed, named nanoprobiotics, which can adhere to inflamed colonic epithelium and eliminate intestinal excess reactive oxygen species in the murine IBD model. Moreover, combined with the anti-inflammatory medicine metformin, nanoprobiotics could further remold the pro-inflammatory microenvironment, improve the overall richness and diversity of the gut microbiota, and exhibit better therapeutic efficacy than commercial IBD chemotherapeutics. Importantly, insignificant overt systemic toxicity in this treatment was observed. By integrating cytokine storm calm with biotherapy, we develop a safe and effective bionanoplatform for the effective treatment of inflammation-mediated intestinal diseases.

6.
Int J Mol Sci ; 25(3)2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38339099

RESUMEN

A cell's ability to secrete extracellular vesicles (EVs) for communication is present in all three domains of life. Notably, Gram-negative bacteria produce a specific type of EVs called outer membrane vesicles (OMVs). We previously observed the presence of OMVs in human blood, which could represent a means of communication from the microbiota to the host. Here, in order to investigate the possible translocation of OMVs from the intestine to other organs, the mouse was used as an animal model after OMVs administration. To achieve this, we first optimized the signal of OMVs containing the fluorescent protein miRFP713 associated with the outer membrane anchoring peptide OmpA by adding biliverdin, a fluorescence cofactor, to the cultures. The miRFP713-expressing OMVs produced in E. coli REL606 strain were then characterized according to their diameter and protein composition. Native- and miRFP713-expressing OMVs were found to produce homogenous populations of vesicles. Finally, in vivo and ex vivo fluorescence imaging was used to monitor the distribution of miRFP713-OMVs in mice in various organs whether by intravenous injection or oral gavage. The relative stability of the fluorescence signals up to 3 days post-injection/gavage paves the way to future studies investigating the OMV-based communication established between the different microbiotas and their host.


Asunto(s)
Escherichia coli , Vesículas Extracelulares , Animales , Ratones , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Distribución Tisular , Vesículas Extracelulares/metabolismo , Intestinos , Bacterias Gramnegativas/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo
7.
Small ; 19(42): e2303225, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37330651

RESUMEN

Cancer vaccines are designed to motivate antigen-specific immune responses and facilitate tumor regression with minimal side effects. To fully exert the potential of vaccines, rationally designed formulations that effectively deliver antigens and trigger potent immune reactions are urgently needed. This study demonstrates a simple and controllable vaccine-developing strategy that assembles tumor antigens into bacterial outer membrane vesicles (OMVs), natural delivery vehicles with intrinsic immune adjuvant properties, via electrostatic interaction. This OMV-delivered vaccine (OMVax) stimulated both innate and adaptive immune responses, leading to enhanced metastasis inhibition and prolonged survival of tumor-bearing mice. Moreover, the influence of different surface charged OMVax on antitumor immunity activation is investigated and declined immune response activation occurred with increased positive surface charge. Together, these findings suggest a simple vaccine formulation that can be enhanced by optimizing the surface charges of vaccine formulations.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Animales , Ratones , Antígenos , Adyuvantes Inmunológicos , Neoplasias/terapia
8.
Appl Environ Microbiol ; 89(3): e0204722, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36809058

RESUMEN

Bacterial outer membrane vesicles (OMVs) are considered a promising vaccine platform for their high built-in adjuvanticity and ability to efficiently induce immune responses. OMVs can be engineered with heterologous antigens based on genetic engineering strategies. However, several critical issues should still be validated, including optimal exposure to the OMV surface, increased production of foreign antigens, nontoxicity, and induction of powerful immune protection. In this study, engineered OMVs with the lipoprotein transport machinery (Lpp) were designed to present SaoA antigen as a vaccine platform against Streptococcus suis. The results suggest that Lpp-SaoA fusions can be delivered on the OMV surface and do not have significant toxicity. Moreover, they can be engineered as lipoprotein and significantly accumulated in OMVs at high levels, thus accounting for nearly 10% of total OMV proteins. Immunization with OMVs containing Lpp-SaoA fusion antigen induced strong specific antibody responses and high levels of cytokines, as well as a balanced Th1/Th2 immune response. Furthermore, the decorated OMV vaccination significantly enhanced microbial clearance in a mouse infection model. It was found that antiserum against lipidated OMVs significantly promoted the opsonophagocytic uptake of S. suis in RAW246.7 macrophages. Lastly, OMVs engineered with Lpp-SaoA induced 100% protection against a challenge with 8× the 50% lethal dose (LD50) of S. suis serotype 2 and 80% protection against a challenge with 16× the LD50 in mice. Altogether, the results of this study provide a promising versatile strategy for the engineering of OMVs and suggest that Lpp-based OMVs may be a universal adjuvant-free vaccine platform for important pathogens. IMPORTANCE Bacterial outer membrane vesicles (OMVs) have become a promising vaccine platform due to their excellent built-in adjuvanticity properties. However, the location and amount of the expression of the heterologous antigen in the OMVs delivered by the genetic engineering strategies should be optimized. In this study, we exploited the lipoprotein transport pathway to engineer OMVs with heterologous antigen. Not only did lapidated heterologous antigen accumulate in the engineered OMV compartment at high levels, but also it was engineered to be delivered on the OMV surface, thus leading to the optimal activation of antigen-specific B cells and T cells. Immunization with engineered OMVs induced a strong antigen-specific antibodies in mice and conferred 100% protection against S. suis challenge. In general, the data of this study provide a versatile strategy for the engineering of OMVs and suggest that OMVs engineered with lipidated heterologous antigens may be a vaccine platform for significant pathogens.


Asunto(s)
Streptococcus suis , Vacunas , Animales , Ratones , Streptococcus suis/genética , Streptococcus suis/metabolismo , Antígenos Heterófilos , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/metabolismo , Lipoproteínas/genética , Anticuerpos Antibacterianos , Vacunas Bacterianas/genética
9.
Int J Mol Sci ; 24(7)2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37047578

RESUMEN

Neutrophil elastase (NE) contributes to innate antibacterial defense at both the intracellular (phagocytosis) and extracellular (degranulation, NETosis) levels. Moraxella catarrhalis, a human respiratory pathogen, can exist in an inflammatory milieu which contains NE. No data are available on the action of NE against M. catarrhalis or on the counteraction of NE-dependent host defenses by this pathogen. Using time-kill assays we found that bacteria are able to survive and replicate in the presence of NE. Transmission electron microscopy and flow cytometry studies with NE-treated bacteria revealed that while NE admittedly destabilizes the outer membrane leaflet, it does not cause cytoplasmic membrane rupture, suggesting that the enzyme does not target components that are essential for cell integrity. Using LC-MS/MS spectroscopy we determined that NE cleaved at least three virulent surface proteins in outer membrane vesicles (OMVs) of M. catarrhalis, including OMP CD, McaP, and TbpA. The cleavage of OMP CD contributes to the significant decrease in resistance to serum complement in the complement-resistant strain Mc6. The cleavage of McaP did not cause any sensitization to erythromycin nor did NE disturb its drug action. Identifying NE as a novel but subtle anti-virulence agent together with its extracellularly not-efficient bactericidal activity against M. catarrhalis may facilitate the pathogen's existence in the airways under inflammation.


Asunto(s)
Elastasa de Leucocito , Moraxella catarrhalis , Humanos , Moraxella catarrhalis/metabolismo , Elastasa de Leucocito/metabolismo , Cromatografía Liquida , Proteínas de la Membrana Bacteriana Externa/metabolismo , Espectrometría de Masas en Tándem , Bacterias/metabolismo
10.
J Proteome Res ; 21(7): 1707-1717, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35674493

RESUMEN

In recent years, the intracellular mechanisms that contribute to antibiotic resistance have received increasing attention, and outer membrane vesicles (OMVs) have been reported to be related to antibiotic resistance in several Gram-negative bacterial species. However, the intrinsic molecular mechanisms and the form of such antibiotic resistance are still largely unknown. In this study, OMVs from an oxytetracycline (OXY) sensitive aquatic pathogen, Aeromonas hydrophila (OXY-S), were found with significantly increased OXY resistance. Interestingly, the OXY-resistant strain (OXY-R) had a more protective role in OXY resistance. Therefore, a DIA-based quantitative proteomics analysis was performed to compare the differential expression of OMV proteins between OXY-R (OMVsR) and OXY-S (OMVsS). The results showed that seven proteins increased and five proteins decreased in OMVsR vs OMVsS. A subsequent antibiotics susceptibility assay showed that the deletion of icd, rpsF, and iscS significantly increased OXY sensitivity. Moreover, the exogenous addition of the crude OMV fractions of overexpressed recombinant proteins in E. coli with rRpsF, rIcd, rIscS, rOmpA, rPepA, rFrdA, and rRplQ demonstrated that these proteins promoted the OXY resistance of A. hydrophila. Overall, our results indicate the important protective role of OMVs in antibiotic resistance in A. hydrophila and provide novel insights on bacterial antibiotic resistance mechanisms.


Asunto(s)
Aeromonas hydrophila , Oxitetraciclina , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Escherichia coli/metabolismo , Oxitetraciclina/metabolismo , Proteómica/métodos
11.
Subcell Biochem ; 97: 101-150, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33779916

RESUMEN

The release of extracellular vesicles (EVs) is a process conserved across the three domains of life. Amongst prokaryotes, EVs produced by Gram-negative bacteria, termed outer membrane vesicles (OMVs), were identified more than 50 years ago and a wealth of literature exists regarding their biogenesis, composition and functions. OMVs have been implicated in benefiting numerous metabolic functions of their parent bacterium. Additionally, OMVs produced by pathogenic bacteria have been reported to contribute to pathology within the disease setting. By contrast, the release of EVs from Gram-positive bacteria, known as membrane vesicles (MVs), has only been widely accepted within the last decade. As such, there is a significant disproportion in knowledge regarding MVs compared to OMVs. Here we provide an overview of the literature regarding bacterial membrane vesicles (BMVs) produced by pathogenic and commensal bacteria. We highlight the mechanisms of BMV biogenesis and their roles in assisting bacterial survival, in addition to discussing their functions in promoting disease pathologies and their potential use as novel therapeutic strategies.


Asunto(s)
Bacterias Gramnegativas , Bacterias Grampositivas , Células Procariotas
12.
Proc Natl Acad Sci U S A ; 116(43): 21780-21788, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31591215

RESUMEN

Bacterial outer membrane vesicles (OMVs) represent an interesting vaccine platform for their built-in adjuvanticity and simplicity of production process. Moreover, OMVs can be decorated with foreign antigens using different synthetic biology approaches. However, the optimal OMV engineering strategy, which should guarantee the OMV compartmentalization of most heterologous antigens in quantities high enough to elicit protective immune responses, remains to be validated. In this work we exploited the lipoprotein transport pathway to engineer OMVs with foreign proteins. Using 5 Staphylococcus aureus protective antigens expressed in Escherichia coli as fusions to a lipoprotein leader sequence, we demonstrated that all 5 antigens accumulated in the vesicular compartment at a concentration ranging from 5 to 20% of total OMV proteins, suggesting that antigen lipidation could be a universal approach for OMV manipulation. Engineered OMVs elicited high, saturating antigen-specific antibody titers when administered to mice in quantities as low as 0.2 µg/dose. Moreover, the expression of lipidated antigens in E. coli BL21(DE3)ΔompAΔmsbBΔpagP was shown to affect the lipopolysaccharide structure, with the result that the TLR4 agonist activity of OMVs was markedly reduced. These results, together with the potent protective activity of engineered OMVs observed in mice challenged with S. aureus Newman strain, makes the 5-combo-OMVs a promising vaccine candidate to be tested in clinics.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Vesículas Extracelulares/inmunología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/inmunología , Animales , Membrana Externa Bacteriana/inmunología , Membrana Externa Bacteriana/metabolismo , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Lipopolisacáridos/inmunología , Ratones , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología
13.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35055181

RESUMEN

The clinical symptoms of shigellosis, a gastrointestinal infection caused by Shigella spp. range from watery diarrhea to fulminant dysentery. Endemic infections, particularly among children in developing countries, represent the majority of clinical cases. The situation is aggravated due to the high mortality rate of shigellosis, the rapid dissemination of multi-resistant Shigella strains and the induction of only serotype-specific immunity. Thus, infection prevention due to vaccination, encompassing as many of the circulating serotypes as possible, has become a topic of interest. However, vaccines have turned out to be ineffective so far. Outer membrane vesicles (OMVs) are promising novel targets for vaccination. OMVs are constitutively secreted by Gram-negative bacteria including Shigella during growth. They are composed of soluble luminal portions and an insoluble membrane and can contain toxins, bioactive periplasmic and cytoplasmic (lipo-) proteins, (phospho-) lipids, nucleic acids and/or lipopolysaccharides. Thus, OMVs play an important role in bacterial cell-cell communication, growth, survival and pathogenesis. Furthermore, they modulate the secretion and transport of biomolecules, the stress response, antibiotic resistance and immune responses of the host. Thus, OMVs serve as novel secretion machinery. Here, we discuss the current literature and highlight the properties of OMVs as potent vaccine candidates because of their immunomodulatory, antigenic and adjuvant properties.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/uso terapéutico , Disentería Bacilar/prevención & control , Shigella/crecimiento & desarrollo , Animales , Vacunas Bacterianas/farmacología , Modelos Animales de Enfermedad , Desarrollo de Medicamentos , Disentería Bacilar/inmunología , Humanos , Viabilidad Microbiana/efectos de los fármacos , Shigella/efectos de los fármacos , Shigella/metabolismo , Vacunación
14.
Int J Mol Sci ; 23(23)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36498912

RESUMEN

Extracellular vesicles (EVs) are critical elements of cell-cell communication. Here, we characterized the outer membrane vesicles (OMVs) released by specific clones of Escherichia coli isolated from the Long-Term Evolution Experiment after 50,000 generations (50K) of adaptation to glucose minimal medium. Compared with their ancestor, the evolved clones produce small OMVs but also larger ones which display variable amounts of both OmpA and LPS. Tracking ancestral, fluorescently labelled OMVs revealed that they fuse with both ancestral- and 50K-evolved cells, albeit in different proportions. We quantified that less than 2% of the cells from one 50K-evolved clone acquired the fluorescence delivered by OMVs from the ancestral strain but that one cell concomitantly fuses with several OMVs. Globally, our results showed that OMV production in E. coli is a phenotype that varies along bacterial evolution and question the contribution of OMVs-mediated interactions in bacterial adaptation.


Asunto(s)
Escherichia coli , Vesículas Extracelulares , Escherichia coli/genética , Proteínas de la Membrana Bacteriana Externa/genética
15.
Int J Mol Sci ; 23(22)2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36430266

RESUMEN

The microbiota constitutes an important part of the holobiont in which extracellular vesicles (EVs) are key players in health, especially regarding inter- and intra-kingdom communications. Analysis of EVs from the red blood cell concentrates of healthy donors revealed variable amounts of OmpA and LPS in 12 of the 14 analyzed samples, providing indirect experimental evidence of the presence of microbiota EVs in human circulating blood in the absence of barrier disruption. To investigate the role of these microbiota EVs, we tracked the fusion of fluorescent Escherichia coli EVs with blood mononuclear cells and showed that, in the circulating blood, these EVs interacted almost exclusively with monocytes. This study demonstrates that bacterial EVs constitute critical elements of the host-microbiota cellular communication. The analysis of bacterial EVs should thus be systematically included in any characterization of human EVs.


Asunto(s)
Vesículas Extracelulares , Microbiota , Humanos , Estado de Salud , Eritrocitos , Monocitos , Escherichia coli
16.
Int J Mol Sci ; 22(3)2021 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-33498269

RESUMEN

Bacterial membrane vesicles (BMVs) are nanoparticles produced by both Gram-negative and Gram-positive bacteria that can function to modulate immunity in the host. Both outer membrane vesicles (OMVs) and membrane vesicles (MVs), which are released by Gram-negative and Gram-positive bacteria, respectively, contain cargo derived from their parent bacterium, including immune stimulating molecules such as proteins, lipids and nucleic acids. Of these, peptidoglycan (PG) and lipopolysaccharide (LPS) are able to activate host innate immune pattern recognition receptors (PRRs), known as NOD-like receptors (NLRs), such as nucleotide-binding oligomerisation domain-containing protein (NOD) 1, NOD2 and NLRP3. NLR activation is a key driver of inflammation in the host, and BMVs derived from both pathogenic and commensal bacteria have been shown to package PG and LPS in order to modulate the host immune response using NLR-dependent mechanisms. Here, we discuss the packaging of immunostimulatory cargo within OMVs and MVs, their detection by NLRs and the cytokines produced by host cells in response to their detection. Additionally, commensal derived BMVs are thought to shape immunity and contribute to homeostasis in the gut, therefore we also highlight the interactions of commensal derived BMVs with NLRs and their roles in limiting inflammatory diseases.


Asunto(s)
Membrana Externa Bacteriana/inmunología , Proteínas NLR/metabolismo , Nanopartículas/química , Adyuvantes Inmunológicos/administración & dosificación , Animales , Membrana Externa Bacteriana/química , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Nanopartículas/metabolismo
17.
Int J Mol Sci ; 21(8)2020 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-32316670

RESUMEN

Gram-negative bacteria have an outer membrane inhibiting the entry of antibiotics. Porins, found within the outer membrane, are involved in regulating the permeability of ß-lactam antibiotics. ß-lactamases are enzymes that are able to inactivate the antibacterial properties of ß-lactam antibiotics. Interestingly, porins and ß-lactamase are found in outer membrane vesicles (OMVs) of ß-lactam-resistant Escherichia coli and may be involved in the survival of susceptible strains of E. coli in the presence of antibiotics, through the hydrolysis of the ß-lactam antibiotic. In this study, OMVs isolated from ß-lactam-resistant E. coli and from mutants, lacking porin or ß-lactamase, were evaluated to establish if the porins or ß-lactamase in OMVs were involved in the degradation of ß-lactam antibiotics. OMVs isolated from E. coli deficient in ß-lactamase did not show any degradation ability against ß-lactam antibiotics, while OMVs lacking OmpC or OmpF showed significantly lower levels of hydrolyzing activity than OMVs from parent E. coli. These data reveal an important role of OMVs in bacterial defense mechanisms demonstrating that the OmpC and OmpF proteins allow permeation of ß-lactam antibiotics into the lumen of OMVs, and antibiotics that enter the OMVs can be degraded by ß-lactamase.


Asunto(s)
Escherichia coli/crecimiento & desarrollo , Porinas/genética , beta-Lactamasas/genética , beta-Lactamas/química , Membrana Externa Bacteriana/metabolismo , Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hidrólisis , Pruebas de Sensibilidad Microbiana , Mutación , Porinas/metabolismo , beta-Lactamasas/metabolismo , beta-Lactamas/farmacología
18.
J Enzyme Inhib Med Chem ; 34(1): 189-195, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30734607

RESUMEN

The genome of Helicobacter pylori encodes for carbonic anhydrases (CAs, EC 4.2.1.1) belonging to the α- and ß-CA classes, which together with urease, have a pivotal role in the acid acclimation of the microorganism within the human stomach. Recently, in the exoproteome of H. pylori, a CA with no indication of the corresponding class was identified. Here, using the protonography and the mass spectrometry, a CA belonging to the α-class was detected in the outer membrane vesicles (OMVs) generated by planktonic and biofilm phenotypes of four H. pylori strains. The amount of this metalloenzyme was higher in the planktonic OMVs (pOMVs) than in the biofilm OMVs (bOMVs). Furthermore, the content of α-CA increases over time in the pOMVs. The identification of the α-CA in pOMVs and bOMVs might shed new light on the role of this enzyme in the colonization, survival, persistence, and pathogenesis of H. pylori.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Anhidrasas Carbónicas/análisis , Anhidrasas Carbónicas/metabolismo , Helicobacter pylori/enzimología , Helicobacter pylori/metabolismo
19.
Mar Drugs ; 17(1)2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30626008

RESUMEN

Shewanella sp. HM13 is a cold-adapted Gram-negative bacterium isolated from the intestine of a horse mackerel. It produces a large amount of outer membrane vesicles (OMVs), which are particles released in the medium where the bacterium is cultured. This strain biosynthesizes a single major cargo protein in the OMVs, a fact that makes Shewanella sp. HM13 a good candidate for the production of extracellular recombinant proteins. Therefore, the structural characterization of the components of the vesicles, such as lipopolysaccharides, takes on a fundamental role for understanding the mechanism of biogenesis of the OMVs and their applications. The aim of this study was to investigate the structure of the oligosaccharide (OS) isolated from Shewanella sp. HM13 cells as the first step for a comparison with that from the vesicles. The lipooligosaccharide (LOS) was isolated from dry cells, purified, and hydrolyzed by alkaline treatment. The obtained OS was analyzed completely, and the composition of fatty acids was obtained by chemical methods. In particular, the OS was investigated in detail by ¹H and 13C NMR spectroscopy and MALDI-TOF mass spectrometry. The oligosaccharide was characterized by the presence of a residue of 8-amino-3,8-dideoxy-manno-oct-2-ulosonic acid (Kdo8N) and of a d,d-heptose, with both residues being identified in other oligosaccharides from Shewanella species.


Asunto(s)
Membrana Celular/química , Lipopolisacáridos/química , Shewanella , Adaptación Fisiológica , Regiones Antárticas , Conformación de Carbohidratos , Frío , Espectroscopía de Resonancia Magnética
20.
Int J Mol Sci ; 20(22)2019 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-31717311

RESUMEN

The virulence of bacterial outer membrane vesicles (OMVs) contributes to innate microbial defense. Limited data report their role in interspecies reactions. There are no data about the relevance of OMVs in bacterial-yeast communication. We hypothesized that model Moraxella catarrhalis OMVs may orchestrate the susceptibility of pathogenic bacteria and yeasts to cationic peptides (polymyxin B) and serum complement. Using growth kinetic curve and time-kill assay we found that OMVs protect Candida albicans against polymyxin B-dependent fungicidal action in combination with fluconazole. We showed that OMVs preserve the virulent filamentous phenotype of yeasts in the presence of both antifungal drugs. We demonstrated that bacteria including Haemophilus influenza, Acinetobacter baumannii, and Pseudomonas aeruginosa coincubated with OMVs are protected against membrane targeting agents. The high susceptibility of OMV-associated bacteria to polymyxin B excluded the direct way of protection, suggesting rather the fusion mechanisms. High-performance liquid chromatography-ultraviolet spectroscopy (HPLC-UV) and zeta-potential measurement revealed a high sequestration capacity (up to 95%) of OMVs against model cationic peptide accompanied by an increase in surface electrical charge. We presented the first experimental evidence that bacterial OMVs by sequestering of cationic peptides may protect pathogenic yeast against combined action of antifungal drugs. Our findings identify OMVs as important inter-kingdom players.


Asunto(s)
Bacterias/patogenicidad , Membrana Celular/metabolismo , Proteínas del Sistema Complemento/farmacología , Vesículas Extracelulares/metabolismo , Péptidos/farmacología , Suero/metabolismo , Levaduras/patogenicidad , Bacterias/efectos de los fármacos , Bacterias/crecimiento & desarrollo , Cationes , Membrana Celular/efectos de los fármacos , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular/efectos de los fármacos , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/ultraestructura , Fluconazol/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Moraxella/metabolismo , Polimixina B/farmacología , Electricidad Estática , Virulencia/efectos de los fármacos , Levaduras/efectos de los fármacos , Levaduras/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA